Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Mol Psychiatry ; 28(11): 4655-4665, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37730843

RESUMO

Social hierarchy has a profound impact on social behavior, reward processing, and mental health. Moreover, lower social rank can lead to chronic stress and often more serious problems such as bullying victims of abuse, suicide, or attack to society. However, its underlying mechanisms, particularly their association with glial factors, are largely unknown. In this study, we report that astrocyte-derived amphiregulin plays a critical role in the determination of hierarchical ranks. We found that astrocytes-secreted amphiregulin is directly regulated by cAMP response element-binding (CREB)-regulated transcription coactivator 3 (CRTC3) and CREB. Mice with systemic and astrocyte-specific CRTC3 deficiency exhibited a lower social rank with reduced functional connectivity between the prefrontal cortex, a major social hierarchy center, and the parietal cortex. However, this effect was reversed by astrocyte-specific induction of amphiregulin expression, and the epidermal growth factor domain was critical for this action of amphiregulin. These results provide evidence of the involvement of novel glial factors in the regulation of social dominance and may shed light on the clinical application of amphiregulin in the treatment of various psychiatric disorders.


Assuntos
Transdução de Sinais , Fatores de Transcrição , Animais , Camundongos , Anfirregulina/genética , Camundongos Knockout , Predomínio Social , Fatores de Transcrição/metabolismo
2.
Int J Neurosci ; 133(12): 1394-1398, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35603448

RESUMO

PURPOSE/AIM OF THE STUDY: Accumulation of hyperphosphorylated tau is a key pathological finding of Alzheimer's disease. Recently, acetylation of tau is emerging as another key pathogenic modification, especially regarding the acetylation of tau at K280 of the hexapeptide 275VQIINK280, a critical sequence in driving tau aggregation. However, the relationship between these two key post-translational modifications is not well known. In this study, effect of acetylation of tau at K280 on tau phosphorylation profile was investigated. MATERIALS AND METHODS: The human neuroblastoma cell line, SH-SY5Y, was transfected with p300 acetyltransferase and tau to induce acetylation of tau. Phosphorylation profile after acetylation was evaluated on western blot. K280A-mutant tau was transfected to investigate the effect of acetylation of tau at K280 on tau phosphorylation profile. RESULTS: Overexpression of p300 acetyltransferase in tau-transfected SH-SY5Y human neuroblastoma cells increased acetylation of tau. Meanwhile, tau and its phosphorylation also increased at various sites such as S199/202, S202/T205, T231, and S422, but not at S396. However, blocking acetylation only at K280 with K280A-mutant tau reversed the increased phosphorylation of tau at S202/T205, T231, and S422, but not at S199/202 or S396. CONCLUSION: Here we identified tau phosphorylation profile in the context of p300-induced acetylation and K280A-mutant tau, demonstrating that tau acetylation affects phosphorylation differently by residues and that acetylation at K280 is a determinant of phosphorylation at some residues in the context of pathologic acetyltransferase activity. Yet, our results suggest there is a complex interplay yet to be explored between tau acetylation with tau phosphorylation.


Assuntos
Doença de Alzheimer , Neuroblastoma , Humanos , Fosforilação , Proteínas tau/metabolismo , Acetilação , Processamento de Proteína Pós-Traducional , Doença de Alzheimer/metabolismo
3.
Biochem Biophys Res Commun ; 524(3): 764-771, 2020 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-32037089

RESUMO

ß-Amyloid (Aß) plaque in the brains of patients with Alzheimer's disease (AD) is mainly caused by impaired clearance of Aß by glial cells, including microglia and astrocytes. Because microglia play an important protective role in the central nervous system, many efforts have been made to identify agents that effectively improve microglial Aß phagocytosis. This study found that TLQP-21, which is cleaved from VGF (VGF nerve growth factor inducible) precursor protein, enhanced Aß phagocytosis and degradation by microglial BV2 cells. TLQP-21 also improved microglial phagocytic activity and promoted fibrillar amyloid-ß (fAß) uptake by microglial BV2 cells via a C3AR1-dependent mechanism. Moreover, TLQP-21 stimulated Aß degradation by enhancing lysosome activity, thereby enhancing fAß clearance. These results suggest that treatment with TLQP-21 may be a novel therapeutic strategy to efficiently enhance microglial Aß clearance in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Espaço Extracelular/metabolismo , Microglia/metabolismo , Fragmentos de Peptídeos/farmacologia , Amiloide/efeitos dos fármacos , Animais , Linhagem Celular , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos , Microglia/efeitos dos fármacos , Neuropeptídeos/farmacologia , Fagocitose/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Receptores de Complemento/metabolismo
4.
Mol Cell Neurosci ; 95: 13-24, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30594669

RESUMO

The number of neurofibrillary tangles containing abnormal hyperphosphorylated tau protein correlates with the degree of dementia in Alzheimer's disease (AD). In addition, autophagosome accumulation and disturbance of autophagy, the process by which toxic aggregate proteins are degraded in the cytosol, are also found in AD models. These indicate that regulation of the autophagy-lysosome system may be a potential therapeutic target for AD. Activation of transcription factor EB (TFEB), a master regulator of autophagy-lysosome system gene transcription, reduces the amount of tau in APP mice. Here, to identify potential therapeutic compounds for AD, we performed two types of screening to determine pharmacologically active compounds that increase 1) neuronal viability in okadaic acid-induced tau hyperphosphorylation-related neurodegeneration models and 2) nuclear localization of TFEB in high-contents screening. Ouabain, a cardiac glycoside, was discovered as a common hit compound in both screenings. It also exhibited a significant protective effect in tau transgenic fly and mouse models in vivo. This work demonstrates that ouabain enhances activation of TFEB through inhibition of the mTOR pathway and induces downstream autophagy-lysosomal gene expression and cellular restorative properties. Therefore, therapeutic approaches using ouabain reduce the accumulation of abnormal toxic tau in vitro and in vivo.


Assuntos
Doença de Alzheimer/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Ouabaína/farmacologia , Animais , Células Cultivadas , Drosophila melanogaster , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
5.
Neurobiol Dis ; 130: 104519, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31233882

RESUMO

The intraneuronal aggregates of hyperphosphorylated and misfolded tau (neurofibrillary tangles, NFTs) cause a stereotypical spatiotemporal Alzheimer's disease (AD) progression that correlates with the severity of the associated cognitive decline. Kinase activity contributes to the balance between neuron survival and cell death. Hyperactivation of kinases including the conventional protein kinase C (PKC) is a defective molecular event accompanying associative memory loss, tau phosphorylation, and progression of AD or related neurodegenerative diseases. Here, we investigated the ability of small therapeutic compounds (a custom library) to improve tau-induced rough-eye phenotype in a Drosophila melanogaster model of frontotemporal dementia. We also assessed the tau phosphorylation in vivo and selected hit compounds. Among the potential hits, we investigated Ro 31-8220, described earlier as a potent PKCα inhibitor. Ro 31-8220 robustly improved the rough-eye phenotype, reduced phosphorylated tau species in vitro and in vivo, reversed tau-induced memory impairment, and improved the fly motor functions. In a human neuroblastoma cell line, Ro 31-8220 reduced the PKC activity and the tau phosphorylation pattern, but we also have to acknowledge the compound's wide range of biological activity. Nevertheless, Ro 31-8220 is a novel therapeutic mitigator of tau-induced neurotoxocity.


Assuntos
Demência Frontotemporal/metabolismo , Indóis/farmacologia , Emaranhados Neurofibrilares/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas tau/metabolismo , Animais , Modelos Animais de Doenças , Drosophila melanogaster , Avaliação Pré-Clínica de Medicamentos , Emaranhados Neurofibrilares/metabolismo , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos
6.
Neurobiol Dis ; 129: 182-194, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31121321

RESUMO

The link between Val232Met variant of phospholipase D3 (PLD3) and late-onset Alzheimer's disease (AD) is still obscure. While it may not affect directly the amyloid precursor protein function, PLD3 could be regulating multiple cellular compartments. Here, we investigated the function of wild-type human PLD3 (PLD3WT) and the Val232Met variant (PLD3VM) in the presence of ß-amyloid (Aß) in a Drosophila melanogaster model of AD. We expressed PLD3WT in CNS of the Aß-model flies and monitored its effect on the ER stress, cell apoptosis and recovery the Aß-induced cognitive impairment. The expression reduced ER stress and neuronal apoptosis, which resulted in normalized antioxidative phospholipids levels and brain protection. A specific O-glycosylation at pT271 in PLD3 is essential for its normal trafficking and cellular localization. The V232 M substitution impairs this O-glycosylation, leading to enlarged lysosomes and plausibly aberrant protein recycling. PLD3VM was less neuroprotective, and while, PLD3WT expression enhances the lysosomal functions, V232 M attenuated PLD3's trafficking to the lysosomes. Thus, the V232 M mutation may affect AD pathogenesis. Further understanding of the mechanistic role of PLD3 in AD could lead to developing novel therapeutic agents.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Neuroproteção/fisiologia , Fosfolipase D/genética , Fosfolipase D/metabolismo , Animais , Animais Geneticamente Modificados , Drosophila melanogaster , Predisposição Genética para Doença , Glicosilação , Humanos , Mutação , Neurônios/metabolismo , Neurônios/patologia , Transporte Proteico
7.
Traffic ; 16(5): 510-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25615530

RESUMO

The triggering receptor expressed on myeloid cells 2 (TREM2) is an immune-modulatory receptor involved in phagocytosis and inflammation. Mutations of Q33X, Y38C and T66M cause Nasu-Hakola disease (NHD) which is characterized by early onset of dementia and bone cysts. A recent, genome-wide association study also revealed that single nucleotide polymorphism of TREM2, such as R47H, increased the risk of Alzheimer's disease (AD) similar to ApoE4. However, how these mutations affect the trafficking of TREM2, which may affect the normal functions of TREM2, was not known. In this study, we show that TREM2 with NHD mutations are impaired in the glycosylation with complex oligosaccharides in the Golgi apparatus, in the trafficking to plasma membrane and further processing by γ-secretase. Although R47H mutation in AD affected the glycosylation and normal trafficking of TREM2 less, the detailed pattern of glycosylated TREM2 differs from that of the wild type, thus suggesting that precise regulation of TREM2 glycosylation is impaired when arginine at 47 is mutated to histidine. Our results suggest that the impaired glycosylation and trafficking of TREM2 from endoplasmic reticulum/Golgi to plasma membrane by mutations may inhibit its normal functions in the plasma membrane, which may contribute to the disease.


Assuntos
Doença de Alzheimer/metabolismo , Complexo de Golgi/metabolismo , Lipodistrofia/metabolismo , Glicoproteínas de Membrana/genética , Mutação , Oligossacarídeos/metabolismo , Osteocondrodisplasias/metabolismo , Receptores Imunológicos/genética , Panencefalite Esclerosante Subaguda/metabolismo , Doença de Alzheimer/genética , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Glicosilação , Células HeLa , Humanos , Lipodistrofia/genética , Glicoproteínas de Membrana/metabolismo , Osteocondrodisplasias/genética , Polimorfismo de Nucleotídeo Único , Transporte Proteico , Receptores Imunológicos/metabolismo , Panencefalite Esclerosante Subaguda/genética
8.
Ann Neurol ; 75(1): 88-97, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24114864

RESUMO

OBJECTIVE: ß-amyloid plaque is a critical pathological feature of Alzheimer disease. Pathologic studies suggest that neurodegeneration may occur in a retrograde fashion from axon terminals near ß-amyloid plaques, and that plaque may spread through brain regions. However, there is no direct experimental evidence to show transmission of ß-amyloid. METHODS: Microscopic imaging data of ß-amyloid transmission was acquired in cortical neuron cultures from Sprague-Dawley rat embryos using polydimethylsiloxane (PDMS) microfluidic culture chambers and in brain sections from in vivo ß-amyloid injection. RESULTS: We present direct imaging evidence in cultured cortical neurons, using PDMS microfluidic culture chambers, that ß-amyloid is readily absorbed by axonal processes and retrogradely transported to neuronal cell bodies. Transmission of ß-amyloid via neuronal connections was also confirmed in mouse brain. ß-Amyloid absorbed by distal axons accumulates in axonal swellings, mitochondria, and lysosomes of the cell bodies. Interestingly, dynasore, an inhibitor of dynamin, which is a protein indispensable for endocytosis, did not prevent retrograde transport of ß-amyloid, indicating that ß-amyloid is absorbed onto axonal membranes and transmitted via them to the cell body. Dynasore did decrease the transneuronal transmission of ß-amyloid, suggesting that this requires the internalization and secretion of ß-amyloid. INTERPRETATION: Our findings provide direct in vitro and in vivo evidence for spreading of ß-amyloid through neuronal connections, and suggest possible therapeutic approaches to blocking this spread.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Axônios/metabolismo , Membrana Celular/metabolismo , Córtex Cerebral/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Axônios/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Dimetilpolisiloxanos/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Cultura Primária de Células , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley
9.
Cell Biosci ; 13(1): 181, 2023 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-37773139

RESUMO

BACKGROUNDS: The expression of major histocompatibility complex I (MHC-I) in neurons has recently been shown to regulate neurite outgrowth and synaptic plasticity. However, its contribution to neurodegenerative diseases such as Alzheimer's disease (AD) remains largely unknown. METHODS: In this study, we investigated the relationship between impaired MHC-I-ß2M complex and AD in vitro and human AD samples. Interaction between protein was identified by liquid chromatography-tandem mass spectrometry and confirmed by immunoprecipitation. Single-chain trimer of MHC-I-ß2M was generated to study the effect of stabilization of MHC-I-ß2M complex on NCAM1 signaling. RESULTS: MHC-I is destabilized in the brains of AD patients and neuronal cells treated with oligomeric ß-amyloid (Aß). Specifically, Aß oligomers disassemble the MHC-I-ß2-microglobulin (ß2M) complex, leading to reduced interactions with neural cell adhesion molecule 1 (NCAM1), a novel interactor of neuronal MHC-I, and decreased signaling. Inhibition of MHC-I-ß2M complex destabilization by non-dissociable MHC-I-ß2M-peptide complex restored MHC-I-NCAM1 signaling in neuronal cells. CONCLUSIONS: The current study demonstrated that disruption of MHC-1-NCAM1 signaling by Aß induced disassembly of MHC-I-ß2M complex is involved in the pathophysiology of AD. Moreover, our findings suggest modulation of MHC-I stability may be a potential therapeutic target for restoring synaptic function in AD.

10.
J Clin Invest ; 133(8)2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-36917188

RESUMO

The spatiotemporal pattern of the spread of pathologically modified tau through brain regions in Alzheimer's disease (AD) can be explained by prion-like cell-to-cell seeding and propagation of misfolded tau aggregates. Hence, to develop targeted therapeutic antibodies, it is important to identify the seeding- and propagation-competent tau species. The hexapeptide 275VQIINK280 of tau is a critical region for tau aggregation, and K280 is acetylated in various tauopathies, including AD. However, the mechanism that links tau acetylated on lysine 280 (tau-acK280) to subsequent progression to neurodegenerative disease remains unclear. Here, we demonstrate that tau-acK280 is critical for tau propagation processes including secretion, aggregation, and seeding. We developed an antibody, Y01, that specifically targets tau-acK280 and solved the crystal structure of Y01 in complex with an acK280 peptide. The structure confirmed that Y01 directly recognizes acK280 and the surrounding residues. Strikingly, upon interaction with acetylated tau aggregates, Y01 prevented tauopathy progression and increased neuronal viability in neuron cultures and in tau-Tg mice through antibody-mediated neutralization and phagocytosis, respectively. Based on our observations that tau-acK280 is a core species involved in seeding and propagation activities, the Y01 antibody that specifically recognizes acK280 represents a promising therapeutic candidate for AD and other neurodegenerative diseases associated with tauopathy.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Tauopatias , Camundongos , Animais , Anticorpos Monoclonais/farmacologia , Proteínas tau/genética , Proteínas tau/metabolismo , Lisina , Tauopatias/tratamento farmacológico , Modelos Animais de Doenças , Encéfalo/metabolismo
11.
Neurobiol Dis ; 46(1): 52-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22245388

RESUMO

Increasing evidence supports an association between Alzheimer's disease (AD) and diabetes. In this context, anti-diabetic agents such as rosiglitazone and glucagon-like peptide (GLP)-1 have been reported to reduce pathologies associated with AD, including tau hyperphosphorylation, suggesting that such agents might be used to treat AD. One such anti-diabetic agent is sitagliptin, which acts through inhibition of dipeptidyl peptidase (DPP)-IV to increase GLP-1 levels. Given this action, sitagliptin would be predicted to reduce AD pathology. Accordingly, we investigated whether sitagliptin is effective in attenuating AD pathologies, focusing on tau phosphorylation in the OLETF type 2 diabetic rat model. Unexpectedly, we found that sitagliptin was not effective against pathological tau phosphorylation in the hippocampus of OLETF type 2 diabetes rats, and instead aggravated it. This paradoxically increased tau phosphorylation was attributed to activation of the tau kinase, GSK3ß (glycogen synthase kinase 3ß). Sitagliptin also increased ser-616 phosphorylation of the insulin receptor substrate (IRS)-1, suggesting increased insulin resistance in the brain. These phenomena were recapitulated in primary rat cortical neurons treated with sitagliptin, further confirming sitagliptin's effects on AD-related pathologies in neurons. These results highlight the need for caution in considering the use of sitagliptin in AD therapy.


Assuntos
Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/metabolismo , Complicações do Diabetes/induzido quimicamente , Complicações do Diabetes/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Pirazinas/toxicidade , Triazóis/toxicidade , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Animais , Complicações do Diabetes/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/enzimologia , Inibidores da Dipeptidil Peptidase IV/toxicidade , Hipocampo/efeitos dos fármacos , Hipocampo/enzimologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/patologia , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Ratos , Ratos Endogâmicos OLETF , Ratos Long-Evans , Fosfato de Sitagliptina
12.
Neurobiol Dis ; 40(2): 449-55, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20655383

RESUMO

Increasing evidence supports an association between Alzheimer's disease (AD) and diabetes. Rosiglitazone, a peroxisome proliferator-activated receptor-γ (PPARγ) agonist, which is an anti-diabetic agent against type 2 diabetes, is currently in Phase III clinical trials in AD patients because rosiglitazone reduces ß-amyloid (Aß) pathology and inflammation. However, few studies have investigated whether rosiglitazone affects tau phosphorylation, another critical pathological feature of AD. Thus, we investigated it using OLETF type 2 diabetic rats and streptozotocin-injected type 1 diabetic mice. Interestingly, rosiglitazone reduced tau phosphorylation only in the hippocampus of OLETF type 2 diabetes rats, and not in that of STZ-injected type 1 diabetes mice. The activity of JNK was reduced in the hippocampus of rosiglitazone-treated OLETF rats, correlating with a reduction in tau phosphorylation, however, which was not correlated with GSK3ß activity. In human tau-transfected SH-SY5Y neuronal cell line, reduction of tau phosphorylation was also associated with reduction of JNK activity, not of GSK3ß activity. Hence, rosiglitazone could be used in reducing tau phosphorylation through JNK inactivation for therapeutic effects in type 2 diabetes related Alzheimer's disease.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipocampo/metabolismo , Hipoglicemiantes/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neurônios/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Proteínas tau/metabolismo , Animais , Western Blotting , Linhagem Celular , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hipocampo/efeitos dos fármacos , Humanos , Lactente , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/metabolismo , PPAR gama/agonistas , Fosforilação , Reação em Cadeia da Polimerase , Ratos , Ratos Endogâmicos OLETF , Rosiglitazona , Transfecção/métodos , Resultado do Tratamento
13.
Biochem Biophys Res Commun ; 395(2): 207-12, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20362550

RESUMO

Glycogen synthase kinase-3beta (GSK3beta) is recognized as one of major kinases to phosphorylate tau in Alzheimer's disease (AD), thus lots of AD drug discoveries target GSK3beta. However, the inactive form of GSK3beta which is phosphorylated at serine-9 is increased in AD brains. This is also inconsistent with phosphorylation status of other GSK3beta substrates, such as beta-catenin and collapsin response mediator protein-2 (CRMP2) since their phosphorylation is all increased in AD brains. Thus, we addressed this paradoxical condition of AD in rat neurons treated with okadaic acid (OA) which inhibits protein phosphatase-2A (PP2A) and induces tau hyperphosphorylation and cell death. Interestingly, OA also induces phosphorylation of GSK3beta at serine-9 and other substrates including tau, beta-catenin and CRMP2 like in AD brains. In this context, we observed that GSK3beta inhibitors such as lithium chloride and 6-bromoindirubin-3'-monoxime (6-BIO) reversed those phosphorylation events and protected neurons. These data suggest that GSK3beta may still have its kinase activity despite increase of its phosphorylation at serine-9 in AD brains at least in PP2A-compromised conditions and that GSK3beta inhibitors could be a valuable drug candidate in AD.


Assuntos
Doença de Alzheimer/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Neurônios/enzimologia , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/tratamento farmacológico , Animais , Células Cultivadas , Modelos Animais de Doenças , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Indóis/farmacologia , Indóis/uso terapêutico , Cloreto de Lítio/farmacologia , Cloreto de Lítio/uso terapêutico , Ácido Okadáico/toxicidade , Oximas/farmacologia , Oximas/uso terapêutico , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteína Fosfatase 2/antagonistas & inibidores , Ratos , Serina/genética , Serina/metabolismo
14.
Aging Cell ; 19(2): e13078, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31800167

RESUMO

A promising new therapeutic target for the treatment of Alzheimer's disease (AD) is the circadian system. Although patients with AD are known to have abnormal circadian rhythms and suffer sleep disturbances, the role of the molecular clock in regulating amyloid-beta (Aß) pathology is still poorly understood. Here, we explored how the circadian repressors REV-ERBα and ß affected Aß clearance in mouse microglia. We discovered that, at Circadian time 4 (CT4), microglia expressed higher levels of the master clock protein BMAL1 and more rapidly phagocytosed fibrillary Aß1-42 (fAß1-42 ) than at CT12. BMAL1 directly drives transcription of REV-ERB proteins, which are implicated in microglial activation. Interestingly, pharmacological inhibition of REV-ERBs with the small molecule antagonist SR8278 or genetic knockdown of REV-ERBs-accelerated microglial uptake of fAß1-42 and increased transcription of BMAL1. SR8278 also promoted microglia polarization toward a phagocytic M2-like phenotype with increased P2Y12 receptor expression. Finally, constitutive deletion of Rev-erbα in the 5XFAD model of AD decreased amyloid plaque number and size and prevented plaque-associated increases in disease-associated microglia markers including TREM2, CD45, and Clec7a. Altogether, our work suggests a novel strategy for controlling Aß clearance and neuroinflammation by targeting REV-ERBs and provides new insights into the role of REV-ERBs in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Relógios Circadianos/genética , Microglia/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Placa Amiloide/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição ARNTL/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/síntese química , Animais , Proteínas CLOCK/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Isoquinolinas/farmacologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/antagonistas & inibidores , Placa Amiloide/genética , Placa Amiloide/patologia , RNA Interferente Pequeno , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética , Receptores Purinérgicos P2Y12/efeitos dos fármacos , Receptores Purinérgicos P2Y12/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Sinapses/genética , Sinapses/metabolismo , Tiofenos/farmacologia
15.
Mol Pain ; 5: 67, 2009 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19922668

RESUMO

BACKGROUND: Recent studies show that transcriptional activation of GTP cyclohydrolase I (GCH1) in dorsal root ganglia (DRG) is significantly involved in the development and persistency of pain symptoms. We thus hypothesize that neuropathic pain may be attenuated by down-regulation of GCH1 expression, and propose a gene silencing system for this purpose. RESULTS: To interrupt GCH1 synthesis, we designed a bidirectional recombinant adeno-associated virus encoding both a small hairpin RNA against GCH1 and a GFP reporter gene (rAAV-shGCH1). After rAAV-shGCH1 was introduced into the sciatic nerve prior to or following pain-inducing surgery, therapeutic efficacy and the underlying mechanisms were subsequently validated in animal models. The GFP expression data indicates that rAAV effectively delivered transgenes to DRG. Subsequently reduced GCH1 expression was evident from immunohistochemistry and western-blotting analysis. Along with the down-regulation of GCH1, the von Frey test correspondingly indicated a sharp decline in pain symptoms upon both pre- and post-treatment with rAAV-shGCH1. Interestingly, GCH1 down-regulation additionally led to decreased microglial activation in the dorsal horn, implying an association between pain attenuation and reduced inflammation. CONCLUSION: Therefore, the data suggests that GCH1 levels can be reduced by introducing rAAV-shGCH1, leading to pain relief. Based on the results, we propose that GCH1 modulation may be developed as a clinically applicable gene therapy strategy to treat neuropathic pain.


Assuntos
Dependovirus/genética , GTP Cicloidrolase/genética , Terapia Genética/métodos , Neuralgia/terapia , RNA Interferente Pequeno/metabolismo , Animais , Western Blotting , Linhagem Celular , GTP Cicloidrolase/metabolismo , Vetores Genéticos/genética , Células HeLa , Humanos , Imuno-Histoquímica , RNA Interferente Pequeno/genética , Ratos
16.
J Virol ; 82(23): 11976-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18799585

RESUMO

Coxsackievirus is the most important cause of meningitis and encephalitis in infants; an infection is sometimes fatal or may lead to neurodevelopmental defects. Here, we show that coxsackievirus B4 (CVB4) induces an autophagy pathway for replication in rat primary neurons. Notably, calpain inhibitors reduce autophagosome formation. Conversely, the inhibition of the autophagy pathway with 3-methyladenine inhibits calpain activation. This work reveals, for the first time, that calpain is essential for the autophagy pathway and viral replication in CVB4-infected neurons.


Assuntos
Autofagia , Calpaína/fisiologia , Enterovirus Humano B/fisiologia , Neurônios/virologia , Replicação Viral , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Apoptose , Células Cultivadas , Córtex Cerebral/virologia , Ratos
17.
J Chem Inf Model ; 49(11): 2419-28, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19852439

RESUMO

In this report, we present a novel virtual high-throughput screening methodology to assist in computer-aided drug discovery. Our method, designated as SLIM, involves ligand-free shape and chemical feature matching. The procedure takes advantage of a negative image of a binding pocket in a target receptor. The negative image is a set of virtual atoms representing the inner shape and chemical features of the binding pocket. Using this image, SLIM implements a shape-based similarity search based on molecular volume superposition for the ensemble of conformers of each molecule. The superposed structures, prioritized by shape similarity, are subjected to comparison of chemical feature similarities. To validate the merits of the SLIM method, we compared its performance with those of three distinct widely used tools ROCS, GLIDE, and GOLD. ROCS was selected as a representative of the ligand-centric methods, and docking programs GLIDE and GOLD as representatives of the receptor-centric methods. Our data suggest that SLIM has overall hit ranking ability that is comparable to that of the docking method, retaining the high computational speed of the ligand-centric method. It is notable that the SLIM method offers consistently reliable screening quality against conformational variations of receptors, whereas the docking methods have limited screening performance.


Assuntos
Receptores de Superfície Celular/metabolismo , Área Sob a Curva , Sítios de Ligação , Ligantes , Conformação Molecular
18.
J Neurosci Res ; 86(14): 3230-9, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18615642

RESUMO

Autophagosomes are accumulated in Alzheimer's disease (AD), but the regulatory pathway of autophagy in AD remains largely unknown. By using electron microscopy, Western blotting, and immunocytochemistry, here we show that autophagosomes are accumulated in rat neurons by okadaic acid (OA), a protein phosphatase-2A inhibitor known to enhance tau phosphorylation, beta-amyloid (Abeta) deposition, and neuronal death, which are the pathological hallmarks of AD. Autophagy can be generally induced via several distinct pathways, such as inhibition of mTOR or activation of beclin-1. Interestingly, OA increased both mTOR and beclin-1 pathways simultaneously, which suggests that autophagy in OA-treated neurons is induced mainly via the beclin-1 pathway, and less so via mTOR inhibition. Finally, inhibition of autophagy by 3MA reduced cytotoxicity in OA-treated neurons. Our novel findings provide new insights into the pathology of and therapeutic intervention for AD.


Assuntos
Doença de Alzheimer/metabolismo , Inibidores Enzimáticos/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Ácido Okadáico/toxicidade , Fagossomos/efeitos dos fármacos , Adenina/análogos & derivados , Adenina/farmacologia , Doença de Alzheimer/patologia , Animais , Proteínas Reguladoras de Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Proteína Beclina-1 , Western Blotting , Imuno-Histoquímica , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Microscopia Eletrônica de Transmissão , Fármacos Neuroprotetores/farmacologia , Fagossomos/ultraestrutura , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/metabolismo , Ratos , Serina-Treonina Quinases TOR
19.
Neurosci Lett ; 437(2): 111-5, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18448253

RESUMO

Impairment of protein phosphatase 2A (PP2A) activity is implicated in tau hyperphosphorylation and microtubule (MT) instability in Alzheimer's disease (AD). Here, we report that okadaic acid, an effective PP2A inhibitor, suppresses the levels of acetylated and detyrosinated tubulins, but enhances tyrosinated tubulins in rat primary cortical neuron cultures. Immunocytochemistry experiments reveal that MTs accumulate intensely around soma and proximal neurites, implying impairment of MT transport to distal neurites which is mediated by dynein and dynactin. Here, we reveal that they can be cleaved by calpain. Notably, shortening of process length in OA-treated neurons is alleviated when calpain cleavage activity is inhibited. Based on these results, we propose that calpain-mediated dynein cleavage in OA-treated neurons is responsible for the MT transport deficit, and consequently, neurite retraction.


Assuntos
Dineínas/metabolismo , Inibidores Enzimáticos/farmacologia , Microtúbulos/metabolismo , Neurônios/metabolismo , Ácido Okadáico/farmacologia , Acetilação/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Animais , Transporte Biológico/fisiologia , Cloreto de Cálcio , Hidróxido de Cálcio/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Combinação de Medicamentos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Cloreto de Potássio , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Ratos , Bicarbonato de Sódio , Cloreto de Sódio , Tubulina (Proteína)/metabolismo , Tirosina/metabolismo
20.
Neurosci Lett ; 423(1): 82-7, 2007 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-17662525

RESUMO

Although the tissue plasminogen activator (tPA)/plasminogen/plasmin proteolytic system is thought to modulate the catabolism of amyloid-beta (Abeta), in vivo evidence remains insufficient. In the brain of human amyloid precursor protein transgenic Tg2576 mice, we found co-accumulation of tPA and plasminogen at the periphery of compact amyloid deposits, mainly Abeta42-cored plaques, as well as in the walls of blood vessels with cerebral amyloid angiopathy (CAA). This tPA/plasminogen system contained high levels of proteolytic activity. High levels of tPA were also found in reactive astrocytes with increased Abeta42 expression, whereas plasminogen was found only in neurons. When the brain sections of Tg2576 mice were treated with both tPA and plasminogen, levels of thioflavin-S fluorescence, congophilicity and birefringence in the compact amyloid plaques were significantly reduced, and the ultrastructure of Abeta42-fibrils was disrupted. These results suggest that the assembled Abeta42 may promote upregulation of the tPA/plasminogen proteolytic system, which can modulate the deposition of amyloid plaques in vivo.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Placa Amiloide/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Química Encefálica/efeitos dos fármacos , Química Encefálica/genética , Corantes , Vermelho Congo , Corantes Fluorescentes , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo , Plasminogênio/metabolismo , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA